Next Article in Journal
Association between Timing of Epinephrine Administration and Outcomes of Traumatic Out-of-Hospital Cardiac Arrest following Traffic Collisions
Next Article in Special Issue
A Focus on the Role of DSC-PWI Dynamic Radiomics Features in Diagnosis and Outcome Prediction of Ischemic Stroke
Previous Article in Journal
Endoscopic Management of Adenomas in the Ileal Pouch and the Rectal Remnant after Surgical Treatment in Familial Adenomatous Polyposis
Previous Article in Special Issue
Potential of Stroke Imaging Using a New Prototype of Low-Field MRI: A Prospective Direct 0.55 T/1.5 T Scanner Comparison
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Admission Severity of Atrial-Fibrillation-Related Acute Ischemic Stroke in Patients under Anticoagulation Treatment: A Systematic Review and Meta-Analysis

1
Faculty of Medicine and Biomedical Sciences, University of Algarve, 8005-139 Faro, Portugal
2
Algarve Biomedical Center, 8005-139 Algarve, Portugal
3
Stroke Unit, Algarve University Hospital Center, 8000-386 Algarve, Portugal
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(12), 3563; https://doi.org/10.3390/jcm11123563
Submission received: 19 May 2022 / Revised: 8 June 2022 / Accepted: 15 June 2022 / Published: 20 June 2022
(This article belongs to the Special Issue New Advances in Diagnostic Radiology of Ischemic Stroke)

Abstract

:
Background: In non-valvular-associated atrial fibrillation (AF), direct oral anticoagulants (DOAC) are as effective as vitamin K antagonists (VKA) for the prevention of acute ischemic stroke (AIS). DOAC are associated with decreased risk and severity of intracranial hemorrhage. It is unknown if different pre-admission anticoagulants impact the prognosis of AF related AIS (AF-AIS). We sought to analyze the literature to assess the association between pre-admission anticoagulation (VKA or DOAC) and admission severity of AF-AIS. Methods: A Systematic literature search (PubMed and ScienceDirect) between January 2011 to April 2021 was undertaken to identify studies describing the outcome of AF-AIS. Results: A total of 128 articles were identified. Of 9493 patients, 1767 were on DOAC, 919 were on therapeutical VKA, 792 were on non-therapeutical VKA and 6015 were not anticoagulated. In comparison to patients without anticoagulation, patients with therapeutical VKA and under DOAC presented with less severe stroke (MD −1.69; 95% CI [−2.71, −0.66], p = 0.001 and MD −2.96; 95% Cl [−3.75, −2.18], p < 0.00001, respectively). Patients with non-therapeutical VKA presented with more severe stroke (MD 1.28; 95% Cl [0.45, 2.12], p = 0.003). Conclusions: In AF-AIS, patients under therapeutical VKA or DOAC have reduced stroke severity on admission in comparison to patients without any anticoagulation, with higher magnitude of protection for DOAC.

1. Introduction

Acute ischemic stroke (AIS) is a major cause of disability and death worldwide [1]. Atrial fibrillation (AF) is the most common cardioembolic etiology, associated with 15 to 30% of all cases of AIS [2,3]. Without treatment, AF is associated with a five-fold increased risk of AIS [4,5,6]. The burden of AF related AIS (AF-AIS) is well characterized. AF-AIS is associated with higher mortality, greater disability, prolonged hospitalization, and a lower chance of being discharged home [7,8,9,10,11,12,13,14,15]. Oral anticoagulants (OAC), namely vitamin K antagonists (VKA) and direct oral anticoagulants (DOAC), reduce the risk of AIS [4,7,16,17,18]. In non-valvular AF, the efficacy of DOAC parallels that of other OAC. Moreover, in comparison to patients taking VKA, patients under treatment with DOAC have reduced frequency and severity of intracerebral hemorrhage, the most lethal complication of anticoagulation therapy [19].
Reflecting the presence of residual thromboembolic risk, AIS can occur even in patients under adequate OAC. Nevertheless, the annual incidence of AF-AIS in patients without OAC (43.4 per 1000 person-years) is much higher than the 9.4 and 10.4 per 1000 person-years from patients under DOAC and warfarin, respectively [20].
It is unknown if different pre-admission OAC can have different impacts on AF-AIS prognosis. We sought to review the literature to assess the association between pre-admission anticoagulation (VKA or DOAC) therapy and initial severity in patients with AF-AIS.

2. Materials and Methods

This protocol follows the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Protocols (PRISMA-P) guidelines [21,22]. The review was registered within the International Prospective Register of Systematic Reviews on 17 April 2021 (PROSPERO ID: CRD42021249531).

2.1. Literature Research

A systematic literature review was performed using PubMed and ScienceDirect databases. The search strategy in this study was as follows: (“atrial fibrillation”) AND (“prior anticoagulation” OR “prestroke anticoagulation” OR “anticoagulation before stroke” OR “preceding anticoagulation”) AND (“stroke severity” OR “stroke outcome”).

2.2. Data Extraction

Two investigators independently screened articles (title, abstract, full articles) to assess eligibility. Titles, names of authors, year of publication, study design, sample and control group size, study characteristics, outcomes and results were extracted and recorded in a Microsoft Excel spreadsheet. Therapeutical and non-therapeutical dosages of the drugs involved were defined based on the criteria of each author in the respective articles. All duplicated articles were removed. Any disagreement was resolved by a third reviewer.

2.3. Study Selection and Quality Assessment

To assess the impact of preadmission anticoagulation on stroke severity, analysis was limited to studies specifically designed or aimed to compare AF-AIS outcomes between patients taking or not taking any specific OAC. To reduce the risk of information or selection bias as well as socioeconomic status as a confounding bias, studies not specifically designed to compare the outcome with a control group were excluded.
We also excluded articles written in a language other than English. The methodological quality of each study was assessed in a qualitative way. Risk of bias was assessed according to the Cochrane Handbook for Systematic Reviews of Interventions guidelines [23].

2.4. Statistical Analysis

The Review Manager (RevMan) software (version 5.4, Cochrane Collaboration) was used for data analysis. For the meta-analysis, the included outcomes were measured using the same metrics in a minimum of three studies. Stroke severity score on admission, measured by National Institutes of Health Stroke Scale (NIHSS), was analyzed in subgroups (VKA therapeutical, VKA non-therapeutical and DOAC). For continuous variables, we used Hozo et al.’s [24,25] formula to estimate mean and variance values from the median, interquartile range and size of the samples. Results were reported with effect sizes and 95% confidence intervals (CI). For continuous variables, this was determined using the inverse variance random-effects model. For dichotomous variables, CI and odds ratio (OR) were measured using the Mantel–Haenszel random-effects model. The OR and mean differences were represented on a forest plot. Heterogeneity across studies was quantified based on Higgins statistics, considering I2 ≥ 50% evidence of a substantial heterogeneity in our meta-analysis. Studies not reporting AIS stroke severity on admission with NIHSS as a mean or a median were excluded from the meta-analysis. In addition, a random-effects approach was selected to extrapolate results as a small number of studies—not functionally identical (performed by researchers operating independently)—were included. This model considers the existence of differences between the studies that go beyond simple sample variability [26]. We intended to assess publication bias through funnel plot asymmetry [23].

3. Results

3.1. Study Selection

PRISMA flowchart diagram (Figure 1) outlines the selection and inclusion process. The initial search yielded 128 studies. The number of studies after deduplication was 111, and 31 fulfilled criteria for full-text reading. From those, 25 were excluded. Reasons for exclusion were the absence of a control group (n = 9), prior anticoagulation due to conditions other than AF (n = 4), post-stroke intervention (n = 2), absence of data regarding AIS outcome (n = 1) and lack of quantitative data reporting stroke severity (NIHSS) as a mean or a median (n = 9).

3.2. Study Characteristics

Six eligible studies reporting on stroke severity, enrolling a total of 9493 participants, 3478 anticoagulated patients and 6015 controls, were included. Regarding anticoagulated patients, 1767 were on DOAC and 1711 on VKA. From the latter, only 919 were therapeutically anticoagulated, considering data provided by the authors. Baseline characteristics of the participants involved are summarized in Table 1.

3.3. Quality Assessment

The quality assessment of each study is described in Table A1 and Appendix A. We considered that five studies had a low risk of bias and one had a moderate risk. None of the included articles had a high risk of bias. According to the Cochrane Handbook for Systematic Reviews of Interventions, the funnel plot asymmetry can only be used when there is a minimum of 10 studies included in the meta-analysis, otherwise the test has a low power to distinguish chance from real asymmetry [23]. As we have only included 6 studies in our meta-analysis, publication bias was not assessed.

3.4. General Sociodemographic and Risk Factors

As seen in Table 2, the median age (range 71–80) as well the distribution of gender, vascular risk factors and the thrombotic risk (CHADS2 score 2–3) was in general comparable between the studies and between treatment arms (DOAC versus therapeutic VKA versus non-therapeutic VKA versus controls).

3.5. Stroke Severity

Six studies were included in the quantitative analysis (Figure 2). Of these, five studies reported results on VKA therapy [13,27,28,29,30] and four studies reported results on DOAC therapy [12,13,27,28]. In five studies, there was a difference of AIS severity between therapeutical VKA and non-therapeutical VKA [13,27,28,30]. None of the studies presented a comparison of admission severity between patients taking DOAC and VKA.

3.6. VKA Non-Therapeutical

Five studies reported on stroke severity in patients with non-therapeutical VKA. The studies by Meinel et al. [13] (MD 1.00; 95% CI [0.43, 1.57]), Sakamoto et al. [27] (MD 3.00; 95% CI [0.24, 5.76]) and Yamashiro et al. [28] (MD 2.25; 95% CI [0.74, 3.76]) showed that the severity was lower in the group of patients without any OAC compared to patients with non-therapeutical VKA. Hannon et al. [29] and Matsumoto et al. [30] did not find any significant statistical differences between the two groups, with MD 0.13; 95% CI [−2.51, 2.76] and MD −0.25; 95% CI [−3.57, 3.07], respectively.
Overall, compared with control groups (87.87%, n = 5738), patients with non-therapeutical VKA on admission (12.13%, n = 792) had significantly higher NIHSS scores (MD 1.28; 95% Cl [0.45, 2.12], p = 0.003). I2 was 25%, showing a low heterogeneity in this dataset.

3.7. VKA Therapeutical

Therapeutical INR levels were provided in four studies. Hannon et al. [29] and Meinel et al. [13] revealed differences between patients with therapeutical VKA and those without OAC, MD −2.75; 95% CI [−4.99, −0.51] and MD −1.75; 95% CI [−2.15, −1.35], respectively. The other two studies, by Sakamoto et al. [27] and Yamashiro et al. [28], did not show a significant difference between the two groups, MD −2.00; 95% CI [−4.77, 0.77] and MD 1.45; 95% CI [−1.97, 4.87], respectively.
Overall, in AF-AIS, patients with therapeutical VKA (14.04%, n = 919) had significantly lower NIHSS scores (MD −1.69; 95% CI [−2.71; −0.66], p = 0.001) compared to control groups (89.96%, n = 5626), suggesting a protective effect associated with therapeutical VKA. I2 was 28%, showing a low heterogeneity in this dataset.

3.8. DOAC

Four studies reported results about anticoagulation therapy with DOAC. In all studies, DOAC therapy (23.23%, n = 1767) was associated with lower NIHSS scores (MD −2.96; 95% Cl [−3.75, −2.18], p < 0.00001) compared to control groups (76.77%, n = 5841) [12,13,27,28]. I2 was 51%, showing a substantial heterogeneity in this dataset.
The meta-analysis taking into account all subgroups of OAC (VKA non-therapeutical, VKA therapeutical and DOAC) did not show any significant statistical difference between the anticoagulated and control groups (MD −1.05; 95% CI [−2.11, 0.00], p = 0.05). However, I2 was 93%, showing a substantial heterogeneity in this dataset.

4. Discussion

In this study we assessed associations between pre-admission use of OAC and severity of AF-AIS. The main finding of our analysis was that, despite differences in the mechanism of action, prior use of OAC is associated with lower severity of AF-AIS on admission. Clot formation inhibition depends on the international normalized ratio (INR) for VKA and on plasma concentration for DOAC [31]. It is known that patients on VKA [32] or DOAC [33] therapy present with smaller infarct volumes on diffusion-weighted magnetic resonance imaging. There is some evidence showing an increased susceptibility to fibrinolysis in the presence of VKA or DOAC [31,32]. Surprisingly, patients with non-therapeutic INR on admission present with higher AIS severity. One might speculate that suboptimal INR control may reflect a higher burden of medical comorbidities, poor medication compliance (other than anticoagulation), worse health literacy or even later hospital presentation. All these factors may adversely influence stroke outcome [34]. Another factor that might help to explain these results is VKA’s mechanism of action: these drugs inhibit the production of several factors dependent on vitamin K, including protein C and protein S [35]. In subtherapeutic INR, there is an insufficient suppression of coagulation such that it cannot overcome the reduction in protein C anticoagulant activity [27]. As a result, when compared with no treatment, the use of subtherapeutic VKA may result in larger thrombi formation [27]. Although no direct comparison between DOAC use and VKA use was presented, the magnitude of the protection was higher for patients taking DOAC in comparison to patients taking VKA (MD −2.96; 95% Cl [−3.75, −2.18], p < 0.00001 versus MD −1.69; 95% CI [−2.71; −0.66], p = 0.001). Real life meta-analysis to compare VKA to DOAC impact on AF-AIS outcomes would be interesting as cost-effectiveness is an important factor for decision.
There are some limitations that are worthwhile to discuss. Population heterogeneity is a reality as we analyzed observational studies using different patient selection. For instance, the samples were not controlled for pre-hospital delay which can determine the extent of severity at the time of admission to the hospital. There is a discrepancy between the number of controls and anticoagulated patients, and the number of therapeutically anticoagulated patients is considerably small. Standardized blood plasma or serum concentration therapeutic ranges for DOAC have not been established and quantitative test results have not been correlated with clinical outcomes [36]. Assumption of anticoagulation status for the DOAC group was based on interview or medical prescription as blood drug concentration has not been measured. Nevertheless, adequate adherence to DOAC therapy has been shown to have a beneficial effect on stroke severity on admission [28] and concurrent pro-thrombotic risk factors appear to be relevant when AIS occurs despite adequate OAC [37].
Only six studies were suitable for meta-analysis; publication bias was not assessed (funnel plot asymmetry has low power if n < 10) [23]. This means that we cannot exclude that such bias might have affected our results.

5. Conclusions

In conclusion, our systematic review demonstrated that pre-stroke anticoagulation is associated with less severe presentation of AF-AIS with apparent higher protection in patients under DOAC.

Author Contributions

Conceptualization, C.G., M.S., M.A., M.H., M.M. and P.V.; methodology, M.A. and A.M.; validation, M.S. and M.H.; formal analysis, P.V. and A.M.; investigation, C.G., M.S., M.A., M.H., M.M. and P.V.; writing—original draft preparation, C.G., M.S., M.A., M.H., M.M. and P.V.; writing—review and editing, H.N. and A.M.; supervision, A.M. and H.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Algarve Biomedical Center with funds from the Municipio de Loulé, in Algarve, Faro, Portugal.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Risk of bias Assessment.
Table A1. Risk of bias Assessment.
ABCDRisk of Bias
Meinel et al., 2020 [13] LowAConfounding bias
Sakamoto et al., 2017 [27] ModerateBSelection bias
Yamashiro et al., 2018 [28] LowCInformation bias
Hoyer et al., 2018 [12] LowDReporting bias
Hannon et al., 2011 [29] Low
Matsumoto et al., 2017 [30] Low Low
Some concerns
High

References

  1. Yi, X.; Lin, J.; Han, Z.; Luo, H.; Shao, M.; Fan, D.; Qiang, Z. Preceding Antithrombotic Treatment is Associated With Acute Ischemic Stroke Severity and Functional Outcome at 90 Days Among Patients With Atrial Fibrillation. J. Stroke Cerebrovasc. Dis. 2019, 28, 2003–2010. [Google Scholar] [CrossRef] [PubMed]
  2. Jung, Y.H.; Choi, H.Y.; Lee, K.Y.; Cheon, K.; Han, S.W.; Park, J.H.; Cho, H.J.; Park, H.J.; Nam, H.S.; Heo, J.H.; et al. Stroke Severity in Patients on Non-Vitamin K Antagonist Oral Anticoagulants with a Standard or Insufficient Dose. Thromb. Haemost. 2018, 118, 2145–2151. [Google Scholar] [CrossRef]
  3. Hellwig, S.; Grittner, U.; Audebert, H.; Endres, M.; Haeusler, K.G. Non-Vitamin K-dependent oral anticoagulants have a positive impact on ischaemic stroke severity in patients with atrial fibrillation. EP Europace 2018, 20, 569–574. [Google Scholar] [CrossRef] [PubMed]
  4. January, C.T.; Wann, L.S.; Calkins, H.; Chen, L.Y.; Cigarroa, J.E.; Cleveland, J.C.; Ellinor, P.T.; Ezekowitz, M.D.; Field, M.E.; Furie, K.L.; et al. 2019 AHA/ACC/HRS Focused Update of the 2014 AHA/ACC/HRS Guideline for the Management of Patients With Atrial Fibrillation: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. Circulation 2019, 140, e125–e151. [Google Scholar] [CrossRef] [PubMed]
  5. Boursier-Bossy, V.; Zuber, M.; Emmerich, J. Ischemic stroke and non-valvular atrial fibrillation: When to introduce anticoagulant therapy? J. Med. Vasc. 2020, 45, 72–80. [Google Scholar] [CrossRef] [PubMed]
  6. Wolf, P.A.; Abbott, R.D.; Kannel, W.B. Atrial fibrillation as an independent risk factor for stroke: The framingham study. Stroke 1991, 22, 983–988. [Google Scholar] [CrossRef] [Green Version]
  7. Lip, G.Y.; Lane, D.A. Stroke prevention in atrial fibrillation: A systematic review. JAMA J. Am. Med. Assoc. 2015, 313, 1950–1962. [Google Scholar] [CrossRef]
  8. Wodchis, W.P.; Bhatia, R.S.; Leblanc, K.; Meshkat, N.; Morra, D. A review of the cost of atrial fibrillation. Value Health 2012, 15, 240–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Tziomalos, K.; Giampatzis, V.; Bouziana, S.D.; Spanou, M.; Kostaki, S.; Papadopoulou, M.; Dourliou, V.; Sofogianni, A.; Savopoulos, C.; Hatzitolios, A.I. Adequacy of preadmission oral anticoagulation with vitamin K antagonists and ischemic stroke severity and outcome in patients with atrial fibrillation. J. Thromb. Thrombolysis 2015, 41, 336–342. [Google Scholar] [CrossRef] [PubMed]
  10. Tavares, S.F.; Ferreira, I.; Chaves, V.; Flores, L.; Correia, C.; Almeida, J.; Fonseca, L.; Chaves, P.C. Acute Ischemic Stroke Outcome and Preceding Anticoagulation: Direct Oral Anticoagulants Versus Vitamin K Antagonists. J. Stroke Cerebrovasc. Dis. 2020, 29, 104691. [Google Scholar] [CrossRef] [PubMed]
  11. Fernandes, L.; Sargento-Freitas, J.; Milner, J.; Silva, A.; Novo, A.; Gonçalves, T.; Marinho, A.V.; Pego, G.M.; Cunha, L.; António, N. Ischemic stroke in patients previously anticoagulated for non-valvular atrial fibrillation: Why does it happen? Rev. Port. Cardiol. 2019, 38, 117–124. [Google Scholar] [CrossRef] [PubMed]
  12. Hoyer, C.; Filipov, A.; Neumaier-Probst, E.; Szabo, K.; Ebert, A.; Alonso, A. Impact of pre-admission treatment with non-vitamin K oral anticoagulants on stroke severity in patients with acute ischemic stroke. J. Thromb. Thrombolysis 2018, 45, 529–535. [Google Scholar] [CrossRef] [PubMed]
  13. Meinel, T.R.; Branca, M.; Marchis, G.M.; Nedeltchev, K.; Kahles, T.; Bonati, L.; Arnold, M.; Heldner, M.R.; Jung, S.; Carrera, E.; et al. Prior Anticoagulation in Patients with Ischemic Stroke and Atrial Fibrillation. Ann. Neurol. 2021, 89, 42–53. [Google Scholar] [CrossRef] [PubMed]
  14. Seiffge, D.J.; Marchis, G.D.; Koga, M.; Paciaroni, M.; Wilson, D.; Capellari, M.; Md, K.M.; Tsivgoulis, G.; Ambler, G.; Arihiro, S.; et al. Ischemic stroke despite oral anticoagulant therapy in patients with atrial fibrillation. Ann. Neurol. 2020, 87, 677–687. [Google Scholar] [CrossRef] [Green Version]
  15. Xian, Y.; O’Brien, E.C.; Liang, L.; Xu, H.; Schwamm, L.; Fonarow, G.; Bhatt, D.L.; Smith, E.E.; Olson, D.M.; Maisch, L.; et al. Association of preceding antithrombotic treatment with acute ischemic stroke severity and in-hospital outcomes among patients with atrial fibrillation. JAMA J. Am. Med. Assoc. 2017, 317, 1057–1067. [Google Scholar] [CrossRef] [PubMed]
  16. Jame, S.; Barnes, G. Stroke and thromboembolism prevention in atrial fibrillation. Heart 2020, 106, 10–17. [Google Scholar] [CrossRef]
  17. Aguilar, M.I.; Hart, R.; Pearce, L.A. Oral anticoagulants versus antiplatelet therapy for preventing stroke in patients with non-valvular atrial fibrillation and no history of stroke or transient ischemic attacks (Review). Cochrane Database Syst. Rev. 2007, 3, CD006186. [Google Scholar] [CrossRef]
  18. Hindricks, G.; Potpara, T.; Dagres, N.; Arbelo, E.; Bax, J.; Blomström-Lundqvist, C.; Boriani, G.; Castella, M.; Dan, G.A.; Dilaveris, P.E.; et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS). Eur. Heart J. 2021, 42, 373–498. [Google Scholar] [CrossRef]
  19. Adachi, T.; Hoshino, H.; Takagi, M.; Fujioka, S. Volume and Characteristics of Intracerebral Hemorrhage with Direct Oral Anticoagulants in Comparison with Warfarin. Cerebrovasc. Dis. Extra. 2017, 7, 62–71. [Google Scholar] [CrossRef] [Green Version]
  20. Bakhai, A.; Petri, H.; Vahidnia, F.; Wolf, C.; Ding, Y.; Foskett, N.; Sculpher, M. Real-world data on the incidence, mortality, and cost of ischaemic stroke and major bleeding events among non-valvular atrial fibrillation patients in England. J. Eval. Clin. Pract. 2021, 27, 119–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Shamseer, L.; Moher, D.; Clarke, M.; Ghersi, D.; Liberati, A.; Petticrew, M.; Shekelle, P.; Stewart, L.A. Preferred reporting items for systematic review and meta-analysis protocols (prisma-p) 2015: Elaboration and explanation. Br. Med. J. 2015, 349, g7647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Donato, H.; Donato, M. Stages for undertaking a systematic review. Acta Médica Portuguesa. 2019, 32, 227–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Higgins, J.P.T.; Thomas, J.; Chandler, J.; Cumpston, M.; Li, T.; Page, M.J.; Welch, V. Cochrane Handbook for Systematic Reviews of Interventions Version 6.2. Available online: www.training.cochrane.org/handbook (accessed on 6 May 2021).
  24. Hozo, S.P.; Djulbegovic, B.; Hozo, I. Estimating the mean and variance from the median, range, and the size of a sample. BMC Med. Res. Methodol. 2005, 5, 13. [Google Scholar] [CrossRef] [Green Version]
  25. Higgins, J.P.T.; Thompson, S.G. Quantifying heterogeneity in a meta-analysis. Stat. Med. 2002, 21, 1539–1558. [Google Scholar] [CrossRef] [PubMed]
  26. Sousa-Pinto, B.; Azevedo, L. Avaliação Crítica de uma Revisão Sistemática e Meta-Análise: Da Execução da Meta-análise à Exploração da Heterogeneidade. Rev. Soc. Port. Anestesiol. 2019, 28, 187–191. [Google Scholar] [CrossRef]
  27. Sakamoto, Y.; Okubo, S.; Nito, C.; Suda, S.; Matsumoto, N.; Abe, A.; Aoki, J.; Shimoyama, T.; Takayama, Y.; Suzuki, K.; et al. The relationship between stroke severity and prior direct oral anticoagulant therapy in patients with acute ischaemic stroke and non-valvular atrial fibrillation. Eur. J. Neurol. 2017, 24, 1399–1406. [Google Scholar] [CrossRef] [PubMed]
  28. Yamashiro, K.; Kurita, N.; Tanaka, R.; Ueno, Y.; Miyamoto, N.; Hira, K.; Nakajima, S.; Urabe, T.; Hattori, N. Adequate Adherence to Direct Oral Anticoagulant is Associated with Reduced Ischemic Stroke Severity in Patients with Atrial Fibrillation. J. Stroke Cerebrovasc. Dis. 2018, 28, 1773–1780. [Google Scholar] [CrossRef] [PubMed]
  29. Hannon, N.; Callaly, E.; Moore, A.; Chróinín, D.N.; Sheehan, O.; Marnane, M.; Merwick, A.; Kyne, L.; Duggan, J.; McCormack, P.M.E.; et al. Improved late survival and disability after stroke with therapeutic anticoagulation for atrial fibrillation: A population study. Stroke 2011, 42, 2503–2508. [Google Scholar] [CrossRef] [Green Version]
  30. Matsumoto, M.; Sakaguchi, M.; Okazaki, S.; Hashikawa, K.; Takahashi, T.; Matsumoto, M.; Ohtsuki, T.; Shimazu, T.; Yoshimine, T.; Mochizuki, H.; et al. Relationship between infarct volume and prothrombin time-international normalized ratio in ischemic stroke patients with nonvalvular atrial fibrillation. Circ. J. 2017, 81, 391–396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Königsbrügge, O.; Weigel, G.; Quehenberger, P.; Pabinger, I.; Ay, C. Plasma clot formation and clot lysis to compare effects of different anticoagulation treatments on hemostasis in patients with atrial fibrillation. Clin. Exp. Med. 2018, 18, 325–336. [Google Scholar] [CrossRef] [Green Version]
  32. Ay, H.; Arsava, E.M.; Gungor, L.; Greer, D.; Singhal, A.B.; Furie, K.L.; Koroshetz, W.J.; Sorensen, A.G. Admission international normalized ratio and acute infarct volume in ischemic stroke. Ann. Neurol. 2008, 64, 499–506. [Google Scholar] [CrossRef] [PubMed]
  33. Yavasoglu, N.G.; Eren, Y.; Tatar, I.G.; Yalcinkaya, I. Infarct volumes of patients with acute ischemic stroke receiving direct oral anticoagulants due to non-valvular atrial fibrillation. Ann. Indian Acad. Neurol. 2021, 24, 27–31. [Google Scholar] [CrossRef] [PubMed]
  34. Hannon, N.; Arsava, E.M.; Audebert, H.J.; Ay, H.; Crowe, M.; Chróinín, D.N.; Furie, K.; McGorrian, C.; Molshatzki, N.; Murphy, S.; et al. Antithrombotic treatment at onset of stroke with atrial fibrillation, functional outcome, and fatality: A systematic review and meta-analysis. Int. J. Stroke 2015, 10, 808–814. [Google Scholar] [CrossRef] [PubMed]
  35. Takano, K.; Iino, K.; Ibayashi, S.; Tagawa, K.; Sadoshima, S.; Fujishima, M. Hypercoagulable state under low-intensity warfarin anticoagulation assessed with hemostatic markers in cardiac disorders. Am. J. Cardiol. 1994, 74, 935–939. [Google Scholar] [CrossRef]
  36. Chen, A.; Stecker, E.A.; Warden, B. Direct Oral Anticoagulant Use: A Practical Guide to Common Clinical Challenges. J. Am. Heart Assoc. 2020, 9, e017559. [Google Scholar] [CrossRef] [PubMed]
  37. Wańkowicz, P.; Nowacki, P.; Gołąb-Janowska, M. Risk factors for ischemic stroke in patients with non-valvular atrial fibrillation and therapeutic international normalized ratio range. Arch Med. Sci. 2019, 15, 1217–1222. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA-P flowchart of the study inclusion process. PRISMA indicates Preferred Reporting Items for Systematic Reviews and Meta-Analyses.
Figure 1. PRISMA-P flowchart of the study inclusion process. PRISMA indicates Preferred Reporting Items for Systematic Reviews and Meta-Analyses.
Jcm 11 03563 g001
Figure 2. Stroke severity. Forest plot showing the effects of anticoagulation versus control (no anticoagulation) on stroke severity, measured by NIHSS.
Figure 2. Stroke severity. Forest plot showing the effects of anticoagulation versus control (no anticoagulation) on stroke severity, measured by NIHSS.
Jcm 11 03563 g002
Table 1. Description of the studies included in the meta-analysis.
Table 1. Description of the studies included in the meta-analysis.
StudyYearType of StudyInclusion CriteriaExposureOutcomesRisk of Bias
Without OACDOACTherapeutical VKANon-Therapeutical VKA
Meinel et al. [13]2020Observational retrospectiveAIS patients with AF (AF diagnosed either before or after stroke onset) aged ≥18 yearsn = 4964n = 1603INR > 1.7
n = 854
INR < 1.7
n = 604
NIHSS at admissionLow
Sakamoto et al. [27]2017Observational retrospectivePatients with AIS or TIA with known AFn = 352n = 43n = 35n = 54NIHSS at admissionModerate
Yamashiro et al. [28]2018Observational retrospectivePatients with AF who developed AIS or TIAn = 248n = 22INR ≥ 2
n = 16
INR < 2
n = 95
NIHSS at admissionLow
Hoyer et al. [12]2018Observational retrospectivePatients with newly detected AF or known AF admitted for AISn = 277n = 99-NIHSS at admissionLow
Hannon et al. [29]2011Observational prospectivePatients with new stroke events and AF (known or new)n = 62-INR 2–3
n = 14
INR < 2 or > 3
n = 13
NIHSS < 72 hLow
Matsumoto et al. [30]2017Observational retrospectiveAF patients who suffered AISn = 112--n = 26NIHSS at admissionLow
VKA: Vitamin K Antagonists; AIS: Acute Ischemic Stroke; AF: Atrial Fibrillation; DOAC: Direct Oral Anticoagulants; NIHSS: National Institute of Health Stroke Scale; OAC: Oral Anticoagulants; TIA: Transient Ischemic Stroke; IS: Ischemic Stroke.
Table 2. Population and vascular risk factors characteristics in studies included in the meta-analysis.
Table 2. Population and vascular risk factors characteristics in studies included in the meta-analysis.
DOACVKA TherapeuticalVKA Non-TherapeuticalControl Group
Meinel et al. [13]Sakamoto et al. [27]Yamashiro et al. [28]Hoyer et al. [12]Meinel et al. [13]Sakamoto et al. [27]Yamashiro et al. [28]Hannon et al. [29]Meinel et al. [13]Sakamoto et al. [27]Yamashiro et al. [28]Matsumoto et al. [30]Hannon et al. [29]Meinel et al. [13]Sakamoto et al. [27]Yamashiro et al. [28]Matsumoto et al. [30]Hoyer et al. [12]Hannon et al. [29]
Mean age, y79.8--79.280.7---82.1--76-79.4--7679.4-
Median age, y-78
(71–82)
71
(63–81)
--80
(74-88)
80
(73–84)
77
(70–80)
-79
(75–86)
80
(72–85)
-76
(69–80)
-78
(70–85)
74
(67–82)
--77
(66–83)
Sex,
(% female)
46%49%40.6%52.5%44%54%31.3%64.3%54%41%39%45%46%49%42%36.7%49%54.5%50%
Hypertension
(%)
87%74%77.3%86.9%87%69%87.5%71.4%90%67%67.4%86%38.5%82%61%64.9%73%85.6%53%
Diabetes mellitus
(%)
25%26%45.5%28.3%28%17%37.5%21.4%27%20%35.8%24%23.1%21%16%20.2%27%31.8%3.3%
Hyperlipidemia
(%)
66%33%45.5%30.3%67%54%43.8%71.4%64%29%39%41%46.2%60%29%36.7%35%27.1%29.5%
Smokers
(%)
12%5%10.5%1%10%6%28.6%42.9%8%15%25.6%22%61.5%13%17%24.3%30%51%57.9%
CHADS2 score-3
(2–4)
2
(1–3.3)
--3
(2–4)
3
(2.3–4)
--3
(2–4)
2
(2–3)
3.1 ± 1.5--2
(1–3)
2
(1–2)
2.8 ± 1.4--
CG: control group; DOAC: Direct Oral Anticoagulants; tVKA: therapeutical Vitamin K Antagonists; ntVKA: non-therapeutical Vitamin K Antagonists.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Garcia, C.; Silva, M.; Araújo, M.; Henriques, M.; Margarido, M.; Vicente, P.; Nzwalo, H.; Macedo, A. Admission Severity of Atrial-Fibrillation-Related Acute Ischemic Stroke in Patients under Anticoagulation Treatment: A Systematic Review and Meta-Analysis. J. Clin. Med. 2022, 11, 3563. https://doi.org/10.3390/jcm11123563

AMA Style

Garcia C, Silva M, Araújo M, Henriques M, Margarido M, Vicente P, Nzwalo H, Macedo A. Admission Severity of Atrial-Fibrillation-Related Acute Ischemic Stroke in Patients under Anticoagulation Treatment: A Systematic Review and Meta-Analysis. Journal of Clinical Medicine. 2022; 11(12):3563. https://doi.org/10.3390/jcm11123563

Chicago/Turabian Style

Garcia, Catarina, Marcelo Silva, Mariana Araújo, Mariana Henriques, Marta Margarido, Patrícia Vicente, Hipólito Nzwalo, and Ana Macedo. 2022. "Admission Severity of Atrial-Fibrillation-Related Acute Ischemic Stroke in Patients under Anticoagulation Treatment: A Systematic Review and Meta-Analysis" Journal of Clinical Medicine 11, no. 12: 3563. https://doi.org/10.3390/jcm11123563

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop